WO2004024871A2 - Anticorps et methodes de creation d'anticorps genetiquement modifies a forte affinite - Google Patents

Anticorps et methodes de creation d'anticorps genetiquement modifies a forte affinite Download PDF

Info

Publication number
WO2004024871A2
WO2004024871A2 PCT/US2003/028722 US0328722W WO2004024871A2 WO 2004024871 A2 WO2004024871 A2 WO 2004024871A2 US 0328722 W US0328722 W US 0328722W WO 2004024871 A2 WO2004024871 A2 WO 2004024871A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
mmr
cell
gene
antibodies
Prior art date
Application number
PCT/US2003/028722
Other languages
English (en)
Other versions
WO2004024871A3 (fr
Inventor
Luigi Grasso
Original Assignee
Morphotek, Inc.
Nicolaides, Nicholas, E.
Sass, Philip, M.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Morphotek, Inc., Nicolaides, Nicholas, E., Sass, Philip, M. filed Critical Morphotek, Inc.
Priority to CA002544124A priority Critical patent/CA2544124A1/fr
Priority to EP03754530A priority patent/EP1556508A4/fr
Priority to AU2003272352A priority patent/AU2003272352A1/en
Priority to JP2004536226A priority patent/JP2006503035A/ja
Publication of WO2004024871A2 publication Critical patent/WO2004024871A2/fr
Publication of WO2004024871A3 publication Critical patent/WO2004024871A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/01Preparation of mutants without inserting foreign genetic material therein; Screening processes therefor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • C12N15/1024In vivo mutagenesis using high mutation rate "mutator" host strains by inserting genetic material, e.g. encoding an error prone polymerase, disrupting a gene for mismatch repair
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the invention is related to the area of antibody maturation and cellular production. In particular, it is related to the field of mutagenesis.
  • antibodies to block the activity of foreign and/or endogenous polypeptides provides an effective and selective strategy for treating the underlying cause of disease.
  • MAb monoclonal antibodies
  • MAb monoclonal antibodies
  • the FDA approved ReoPro Gaser, V. (1996) Can ReoPro repolish tarnished monoclonal therapeutics? Nat. Biotechnol. 14:1216-1217
  • an anti-platelet MAb from Centocor Herceptin (Weiner, L.M. (1999) Monoclonal antibody therapy of cancer. Semin. Oncol.
  • Standard methods for generating MAbs against candidate protein targets are known by those skilled in the art. Briefly, rodents such as mice or rats are injected with a purified antigen in the presence of adjuvant to generate an immune response (Shield, C.F., et al. (1996) A cost-effective analysis of OKT3 induction therapy in cadaveric kidney transplantation. Am. J. Kidney Dis. 27:855-864). Rodents with positive immune sera are sacrificed and splenocytes are isolated. Isolated splenocytes are fused to melanomas to produce immortalized cell lines that are then screened for antibody production. Positive lines are isolated and characterized for antibody production.
  • rodent MAbs as human therapeutic agents were confounded by the fact that human anti-rodent antibody (HARA) responses occurred in a significant number of patients treated with the rodent- derived antibody (Khazaeli, M.B., et al, (1994) Human immune response to monoclonal antibodies. J. Immunother. 15:42-52).
  • HAA human anti-rodent antibody
  • CDRs complementarity determining regions
  • HAb rodent-derived MAbs
  • Another problem that exists in antibody engineering is the generation of stable, high yielding producer cell lines that is required for manufacturing of the molecule for clinical materials.
  • Several strategies have been adopted in standard practice by those skilled in the art to circumvent this problem.
  • One method is the use of Chinese Hamster Ovary (CHO) cells transfected with exogenous lg fusion genes containing the grafted human light and heavy chains to produce whole antibodies or single chain antibodies, which are a chimeric molecule containing both light and heavy chains that form an antigen-binding polypeptide (Reff, M.E. (1993) High-level production of recombinant immunoglobulins in mammalian cells. Curr. Opin. Biotechnol. 4:573-576).
  • Another method employs the use of human lymphocytes derived from transgenic mice containing a human grafted immune system or transgenic mice containing a human lg gene repertoire.
  • Yet another method employs the use of monkeys to produce primate MAbs, which have been reported to lack a human anti-monkey response (Neuberger, M., and Gruggermann, M. (1997) Monoclonal antibodies. Mice perform a human repertoire. Nature 386:25-26). In all cases, the generation of a cell line that is capable of generating sufficient amounts of high affinity antibody poses a major limitation for producing sufficient materials for clinical studies.
  • a method for generating diverse antibody sequences within the variable domain that results in HAbs and MAbs with high binding affinities to antigens would be useful for the creation of more potent therapeutic and diagnostic reagents respectively.
  • the generation of randomly altered nucleotide and polypeptide residues throughout an entire antibody molecule will result in new reagents that are less antigenic and/or have beneficial pharmacokinetic properties.
  • the invention described herein is directed to the use of random genetic mutation throughout an antibody structure in vivo by blocking the endogenous mismatch repair (MMR) activity of a host cell producing immunoglobulins that encode biochemically active antibodies.
  • MMR mismatch repair
  • the invention also relates to methods for repeated in vivo genetic alterations and selection for antibodies with enhanced binding and pharmacokinetic profiles.
  • the ability to develop genetically altered host cells that are capable of secreting increased amounts of antibody will also provide a valuable method for creating cell hosts for product development.
  • the invention described herein is directed to the creation of genetically altered cell hosts with increased antibody production via the blockade of MMR.
  • the invention facilitates the generation of high affinity antibodies and the production of cell lines with elevated levels of antibody production. Other advantages of the present invention are described in the examples and figures described herein.
  • the invention provides methods for generating genetically altered antibodies (including single chain molecules) and antibody producing cell hosts in vitro and in vivo, whereby the antibody possess a desired biochemical property(s), such as, but not limited to, increased antigen binding, increased gene expression, and/or enhanced extracellular secretion by the cell host.
  • One method for identifying antibodies with increased binding activity or cells with increased antibody production is through the screening of MMR defective antibody producing cell clones that produce molecules with enlianced binding properties or clones that have been genetically altered to produce enhanced amounts of antibody product.
  • the antibody producing cells suitable for use in the invention include, but are not limited to rodent, primate, or human hybridomas or lymphoblastoids; mammalian cells transfected and expressing exogenous lg subunits or chimeric single chain molecules; plant cells, yeast or bacteria transfected and expressing exogenous lg subunits or chimeric single chain molecules.
  • the invention provides methods for making hypermutable antibody-producing cells by introducing a polynucleotide comprising a dominant negative allele of a mismatch repair gene into cells that are capable of producing antibodies.
  • the cells that are capable of producing antibodies include cells that naturally produce antibodies, and cells that are engineered to produce antibodies through the introduction of immunoglobulin encoding sequences.
  • the introduction of polynucleotide sequences into cells is accomplished by transfection.
  • the invention also provides methods of making hypermutable antibody producing cells by introducing a dominant negative mismatch repair (MMR) gene such as PMS2 (preferably human PMS2), MLH1, PMS1, MSH2, or MSH2 into cells that are capable of producing antibodies.
  • MMR dominant negative mismatch repair
  • the dominant negative allele ' of a mismatch repair gene may be a truncation mutation of a mismatch repair gene (preferably a truncation mutation at codon 134, or a thymidine at nucleotide 424 of wild-type PMS2).
  • the invention also provides methods in which mismatch repair gene activity is suppressed. This may be accomplished, for example, using antisense molecules directed against the mismatch repair gene or transcripts.
  • Other embodiments of the invention provide methods for making hypermutable antibody-producing cells by introducing a polynucleotide comprising a dominant negative allele of a mismatch repair gene into fertilized eggs of animals. These methods may also include subsequently implanting the eggs into pseudo-pregnant females whereby the fertilized eggs develop into a mature transgenic animal.
  • the mismatch repair genes may include, for example, PMS2 (preferably human PMS2), MLH1, PMS1, MSH2, or MSH2.
  • the dominant negative allele of a mismatch repair gene may be a truncation mutation of a mismatch repair gene (preferably a truncation mutation at codon 134, or a thymidine at nucleotide 424 of wild-type PMS2).
  • the invention further provides homogeneous compositions of cultured, hypermutable, mammalian cells that are capable of producing antibodies and contain a dominant negative allele of a mismatch repair gene.
  • the mismatch repair genes may include, for example, PMS2 (preferably human PMS2), MLH1, PMS1, MSH2, or MSH2.
  • the dominant negative allele of a mismatch repair gene may be a truncation mutation of a mismatch repair gene (preferably a truncation mutation at codon 134, or a thymidine at nucleotide 424 of wild-type PMS2).
  • the cells of the culture may contain PMS2, (preferably human PMS2), MLH1, or PMS1; or express a human mutL homolog, or the first 133 amino acids of hPMS2.
  • the invention further provides methods for generating a mutation in an immunoglobulin gene of interest by culturing an immunoglobulin producing cell selected for an immunoglobulin of interest wherein the cell contains a dominant negative allele of a mismatch repair gene.
  • the properties of the immunoglobulin produced from the cells can be assayed to ascertain whether the immunoglobulin gene harbors a mutation.
  • the assay may be directed to analyzing a polynucleotide encoding the immunoglobulin, or may be directed to the immunoglobulin polypeptide itself.
  • the invention also provides methods for generating a mutation in a gene affecting antibody production in an antibody-producing cell by culturing the cell expressing a dominant negative allele of a mismatch repair gene, and testing the cell to determine whether the cell harbors mutations within the gene of interest, such that a new biochemical feature (e.g., over-expression and/or secretion of immunoglobulin products) is generated.
  • the testing may include analysis of the steady state expression of the immunoglobulin gene of interest, and/or analysis of the amount of secreted protein encoded by the immunoglobulin gene of interest.
  • the invention also embraces prokaryotic and eukaryotic transgenic cells made by this process, including cells from rodents, non-human primates and humans.
  • aspects of the invention encompass methods of reversibly altering the hypermutability of an antibody producing cell, in which an inducible vector containing a dominant negative allele of a mismatch repair gene operably linked to an inducible promoter is introduced into an antibody-producing cell.
  • the cell is treated with an inducing agent to express the dominant negative mismatch repair gene (which can be PMS2 (preferably human PMS2), MLH1, or PMS1).
  • the cell may be induced to express a human mutL homolog or the first 133 amino acids of hPMS2.
  • the cells may be rendered capable of producing antibodies by co-transfecting a preselected immunoglobulin gene of interest.
  • the immunoglobulin genes of the hypermutable cells, or the proteins produced by these methods may be analyzed for desired properties, and induction may be stopped such that the genetic stability of the host cell is restored.
  • the invention also embraces methods of producing genetically altered antibodies by transfecting a polynucleotide encoding an immunoglobulin protein into a cell containing a dominant negative mismatch repair gene (either naturally or in which the dominant negative mismatch repair gene was introduced into the cell), culturing the cell to allow the immunoglobulin gene to become mutated and produce a mutant immunoglobulin, screening for a desirable property of said mutant immunoglobulin protein, isolating the polynucleotide molecule encoding the selected mutant immunoglobulin possessing the desired property, and transfecting said mutant polynucleotide into a genetically stable cell, such that the mutant antibody is consistently produced without further genetic alteration.
  • the dominant negative mismatch repair gene may be PMS2 (preferably human PMS2), MLH1, or PMS1.
  • the cell may express a human mutL homolog or the first 133 amino acids of hPMS2.
  • the invention further provides methods for generating genetically altered cell lines that express enhanced amounts of an antigen binding polypeptide.
  • antigen-binding polypeptides may be, for example, immunoglobulins.
  • the methods of the invention also include methods for generating genetically altered cell lines that secrete enhanced amounts of an antigen binding polypeptide.
  • the cell lines are rendered hypermutable by dominant negative mismatch repair genes that provide an enhanced rate of genetic hypermutation in a cell producing antigen-binding polypeptides such as antibodies.
  • Such cells include, but are not limited to hybridomas.
  • Expression of enhanced amounts of antigen binding polypeptides may be through enlianced transcription or translation of the polynucleotides encoding the antigen binding polypeptides, or through the enhanced secretion of the antigen binding polypeptides, for example.
  • Methods are also provided for creating genetically altered antibodies in vivo by blocking the MMR activity of the cell host, or by transfecting genes encoding for immunoglobulin in a MMR defective cell host.
  • Antibodies with increased binding properties to an antigen due to genetic changes within the variable domain are provided in methods of the invention that block endogenous MMR of the cell host.
  • Antibodies with increased binding properties to an antigen due to genetic changes within the CDR regions within the light and/or heavy chains are also provided in methods of the invention that block endogenous MMR of the cell host.
  • the invention provides methods of creating genetically altered antibodies in MMR defective Ab producer cell lines with enhanced pharmacokinetic properties in host organisms including but not limited to rodents, primates, and man.
  • a method for making an antibody producing cell line hypermutable is provided.
  • a polynucleotide encoding a dominant negative allele of a MMR gene is introduced into an antibody-producing cell.
  • the cell becomes hypermutable as a result of the introduction of the gene.
  • a method is provided for introducing a mutation into an endogenous gene encoding for an immunoglobulin polypeptide or a single chain antibody.
  • a polynucleotide encoding a dominant negative allele of a MMR gene is introduced into a cell.
  • the cell becomes hypermutable as a result of the introduction and expression of the MMR gene allele.
  • the cell further comprises an immunoglobulin gene of interest.
  • the cell is grown and tested to determine whether the gene encoding for an immunoglobulin or a single chain antibody of interest harbors a mutation.
  • the gene encoding the mutated immunoglobulin polypeptide or single chain antibody may be isolated and expressed in a genetically stable cell.
  • the mutated antibody is screened for at least one desirable property such as, but not limited to, enhanced binding characteristics.
  • a gene or set of genes encoding for lg light and heavy chains or a combination therein are introduced into a mammalian cell host that is MMR defective. The cell is grown, and clones are analyzed for antibodies with enhanced binding characteristics.
  • a method for producing new phenotypes of a cell.
  • a polynucleotide encoding a dominant negative allele of a MMR gene is introduced into a cell.
  • the cell becomes hypermutable as a result of the introduction of the gene.
  • the cell is grown.
  • the cell is tested for the expression of new phenotypes where the phenotype is enhanced secretion of a polypeptide.
  • the invention also provides antibodies having increased affinity for antigen comprising immunoglobulin molecules wherein a substitution has been made for at least one amino acid in the variable domain of the heavy and/or light chain.
  • the substitution is in a position wherein the parental amino acid in that position is an amino acid with a non-polar side chain.
  • the parental amino acid is substituted with a different amino acid that has a non-polar side chain.
  • the parental amino acid is replaced with a proline or hydroxyproline.
  • the substitution(s) are made in the framework regions of the heavy and/or light chain variable domains. In some embodiments, the substitution(s) are made within the first framework region of the heavy chain.
  • the substitution(s) are made within the second framework region of the light chain. In some embodiments, the substitutions are made within the first framework region of the heavy chain and the second framework region of the light chain. In some embodiments, a substitution is made at position 6 of the first framework region of the heavy chain as shown in SEQ ID NO: 18. In some embodiments a substitution is made at position 22 of the second framework region of the light chain as shown in SEQ ID NO:21.
  • the amino acid substitution is a proline or hydroxyproline.
  • the invention also provides methods for increasing the affinity of an antibody for an antigen comprising substituting an amino acid within the variable domain of the heavy or light chain of the subject antibody with another amino acid having a non-polar side chain.
  • a proline is substituted for the original amino acid at the position, h some embodiments, proline is used to substitute for another amino acid having a non-polar side chain.
  • alanine and/or leucine is replaced by proline.
  • the amino acid in position 6 of the first framework region of the heavy chain of the antibody as shown in SEQ ID NO: 18 is replaced with a proline.
  • the amino acid in position 22 of the second framework region of the light chain variable domain as shown in SEQ ID NO:21 is replaced with proline.
  • the invention also provides antibodies produced by these methods.
  • the antibodies produced in the invention may be made using the process of the invention wherein a dominant negative allele of a mismatch repair gene is introduced into the antibody producing cell and the cell becomes hypermutable as described more fully herein.
  • a dominant negative allele of a mismatch repair gene is introduced into the antibody producing cell and the cell becomes hypermutable as described more fully herein.
  • the cells treated with the chemicals that disrupt mismatch repair or which express a dominant-negative mismatch repair gene become hypermutable.
  • the antibodies produced by the hypermutable cells are screened for increased affinity, and those antibodies comprising the amino acid substitutions described above display increased affinity for antigen.
  • the cells producing the antibodies which have the increased affinity and the molecular characteristics described herein may be rendered genetically stable again by withdrawing the chemical inl ibitor, or by rendering the cells genetically stable through the inactivation of the expression of the dominant negative allele.
  • a dominant negative allele that is under the control of an inducible promoter may be inactivated by withdrawing the inducer.
  • the dominant negative allele may be knocked out, or a CRE-LOX expression system may be used whereby the dominant negative allele is spliced from the genome once the cells containing a genetically diverse immunoglobulin have been established.
  • one of skill in the art may use any known method of introducing mutations into proteins and selecting for antibodies having higher affinity with the amino acid substitutions described above. Methods of introducing mutations may be random, such as chemical mutagenesis, or may be specific, such as site-directed mutagenesis.
  • Methods for random and specific mutagenesis include, but are not limited to, for example, chemical mutagenesis (e.g., using such chemicals as methane sulfonate, dimethyl sulfonate, O6-methyl benzadine, methylnitrosourea (MNU), and ethylnitrosourea (ENU)); oligonucleotide-mediated site-directed mutagenesis; alanine scanning; and PCR mutagenesis (see, for example, Kunkel et al. (1991) Methods Enzymol. 204:125-139, site-directed mutagenesis; Crameri et ⁇ /. (1995) BioTechniques 18(2): 194- 196, cassette mutagenesis; and Haught et al. (1994) BioTechniques 16(l):47-48, restriction selection mutagenesis).
  • chemical mutagenesis e.g., using such chemicals as methane sulfonate, dimethyl
  • FIG. 1 Hybridoma cells stably expressing PMS2 and PMS134 MMR genes. Shown is steady state mRNA expression of MMR genes transfected into a murine hybridoma cell line. Stable expression was found after 3 months of continuous growth.
  • the (-) lanes represent negative controls where no reverse transcriptase was added, and the (+) lanes represent samples reverse transcribed and PCR amplified for the MMR genes and an internal housekeeping gene as a control.
  • FIG. 1 Creation of genetically hypermutable hybridoma cells.
  • Dominant negative MMR gene alleles were expressed in cells expressing a MMR-sensitive reporter gene.
  • Dominant negative alleles such as PMS134 and the expression of MMR genes from other species results in antibody producer cells with a hypermutable phenotype that can be used to produce genetically altered immunoglobulin genes with enhanced biochemical features as well as lines with increased lg expression and/or secretion.
  • Values shown represent the amount of converted CPRG substrate which is reflective of the amount of function ⁇ - galactosidase contained within the cell from genetic alterations within the pCAR-OF reporter gene. Higher amounts of ⁇ -galactosidase activity reflect a higher mutation rate due to defective MMR.
  • Figure 3 Screening method for identifying antibody-producing cells containing antibodies with increased binding activity and/or increased expression/secretion.
  • Figure 4. Generation of a genetically altered antibody with an increased binding activity. Shown are ELISA values from 96-well plates, screened for antibodies specific to hlgE. Two clones with a high binding value were found in HB134 cultures.
  • Figure 5. Sequence alteration within variable chain of an antibody (a mutation within the light chain variable region in MMR-defective HB134 antibody producer cells). An arrow indicates the nucleotide at which a mutation occurred in a subset of cells from a clone derived from HB134 cells.
  • Figure 5 A the change results in a Thr to Ser change within the light chain variable region.
  • the coding sequence is in the antisense direction.
  • Figure 5B the change results in a Pro to His change within the light chain variable region.
  • Figure 6. Generation of MMR-defective clones with enhanced steady state lg protein levels.
  • a Western blot of heavy chain immunglobulins from HB134 clones with high levels of MAb (>500ngs/ml) within the conditioned medium shows that a subset of clones express higher steady state levels of immunoglobulins (lg).
  • the H36 cell line was used as a control to measure steady state levels in the parental strain.
  • Lane 1 fibroblast cells (negative control); Lane 2: H36 cell; Lane 3: HB134 clone with elevated MAb levels; Lane 4: HB134 clone with elevated MAb levels; Lane 5: HB134 clone with elevated MAb levels.
  • MMR conserved mismatch repair
  • Blocking MMR in antibody- producing cells such as but not limited to: hybridomas; mammalian cells transfected with genes encoding for lg light and heavy chains; mammalian cells transfected with genes encoding for single chain antibodies; eukaryotic cells transfected with lg genes, can enhance the rate of mutation within these cells leading to clones that have enhanced antibody production and/or cells containing genetically altered antibodies with enhanced biochemical properties such as increased antigen binding.
  • the process of MMR also called mismatch proofreading, is carried out by protein complexes in cells ranging from bacteria to mammalian cells.
  • a MMR gene is a gene that encodes for one of the proteins of such a mismatch repair complex.
  • a MMR complex is believed to detect distortions of the DNA helix resulting from non-complementary pairing of nucleotide bases.
  • the non-complementary base on the newer DNA strand is excised, and the excised base is replaced with the appropriate base, which is complementary to the older DNA strand.
  • cells eliminate many mutations that occur as a result of mistakes in DNA replication.
  • Dominant negative alleles cause a MMR defective phenotype even in the presence of a wild-type allele in the same cell.
  • An example of a dominant negative allele of a MMR gene is the human gene hPMS2-134, which carries a truncating mutation at codon 134 (SEQ ID NO:15). The mutation causes the product of this gene to abnormally terminate at the position of the 134th amino acid, resulting in a shortened polypeptide containing the N-terminal 133 amino acids. Such a mutation causes an increase in the rate of mutations, which accumulate in cells after DNA replication. Expression of a dominant negative allele of a mismatch repair gene results in impairment of mismatch repair activity, even in the presence of the wild-type allele.
  • Dominant negative alleles of a MMR gene can be obtained from the cells of humans, animals, yeast, bacteria, or other organisms. Such alleles can be identified by screening cells for defective MMR activity. Cells from animals or humans with cancer can be screened for defective mismatch repair. Cells from colon cancer patients may be particularly useful. Genomic DNA, cDNA, or mRNA from any cell encoding a MMR protein can be analyzed for variations from the wild type sequence. Dominant negative alleles of a MMR gene can also be created artificially, for example, by producing variants of the hPMS2-134 allele or other MMR genes. Various techniques of site-directed mutagenesis can be used.
  • the suitability of such alleles, whether natural or artificial, for use in generating hypermutable cells or animals can be evaluated by testing the mismatch repair activity caused by the allele in the presence of one or more wild-type alleles, to detennine if it is a dominant negative allele.
  • a cell or an animal into which a dominant negative allele of a mismatch repair gene has been introduced will become hypermutable. This means that the spontaneous mutation rate of such cells or animals is elevated compared to cells or animals without such alleles.
  • the degree of elevation of the spontaneous mutation rate can be at least 2-fold, 5-fold, 10- fold, 20-fold, 50-fold, 100-fold, 200-fold, 500-fold, or 1000-fold that of the normal cell or animal.
  • a polynucleotide encoding for a dominant negative form of a MMR protein is introduced into a cell.
  • the gene can be any dominant negative allele encoding a protein, which is part of a MMR complex, for example, PMS2, PMSl, MLHl, or MSH2.
  • the dominant negative allele can be naturally occurring or made in the laboratory.
  • the polynucleotide can be in the form of genomic DNA, cDNA, RNA, or a chemically synthesized polynucleotide.
  • the polynucleotide can be cloned into an expression vector containing a constitutively active promoter segment (such as but not limited to CMV, SV40, Elongation Factor or LTR sequences) or to inducible promoter sequences such as the steroid inducible pIND vector (Invitrogen), where the expression of the dominant negative MMR gene can be regulated.
  • a constitutively active promoter segment such as but not limited to CMV, SV40, Elongation Factor or LTR sequences
  • inducible promoter sequences such as the steroid inducible pIND vector (Invitrogen)
  • the polynucleotide can be introduced into the cell by transfection.
  • an immunoglobulin (lg) gene, a set of lg genes or a chimeric gene containing whole or parts of an lg gene can be transfected into MMR deficient cell hosts, the cell is grown and screened for clones containing genetically altered lg genes with new biochemical features.
  • MMR defective cells may be of human, primates, mammals, rodent, plant, yeast or of the prokaryotic kingdom.
  • the mutated gene encoding the lg with new biochemical features may be isolated from the respective clones and introduced into genetically stable cells (i.e., cells with normal MMR) to provide clones that consistently produce lg with the new biochemical features.
  • the method of isolating the lg gene encoding lg with new biochemical features may be any method known in the art.
  • Introduction of the isolated polynucleotide encoding the lg with new biochemical features may also be performed using any method known in the art, including, but not limited to transfection of an expression vector containing the polynucleotide encoding the lg with new biochemical features.
  • transfecting an lg gene a set of lg genes or a chimeric gene containing whole or parts of an lg gene into an MMR deficient host cell, such lg genes may be transfected simultaneously with a gene encoding a dominant negative mismatch repair gene into a genetically stable cell to render the cell hypermutable.
  • Transfection is any process whereby a polynucleotide is introduced into a cell.
  • the process of transfection can be carried out in a living animal, e.g., using a vector for gene therapy, or it can be carried out in vitro, e.g., using a suspension of one or more isolated cells in culture.
  • the cell can be any type of eukaryotic cell, including, for example, cells isolated from humans or other primates, mammals or other vertebrates, invertebrates, and single celled organisms such as protozoa, yeast, or bacteria.
  • transfection will be carried out using a suspension of cells, or a single cell, but other methods can also be applied as long as a sufficient fraction of the treated cells or tissue incorporates the polynucleotide so as to allow transfected cells to be grown and utilized.
  • the protein product of the polynucleotide may be transiently or stably expressed in the cell.
  • Techniques for transfection are well known. Available techniques for introducing polynucleotides include but are not limited to electroporation, transduction, cell fusion, the use of calcium chloride, and packaging of the polynucleotide together with lipid for fusion with the cells of interest.
  • An isolated cell is a cell obtained from a tissue of humans or animals by mechanically separating out individual cells and transferring them to a suitable cell culture medium, either with or without pretreatment of the tissue with enzymes, e.g., collagenase or trypsin. Such isolated cells are typically cultured in the absence of other types of cells.
  • Cells selected for the introduction of a dominant negative allele of a mismatch repair gene may be derived from a eukaryotic organism in the form of a primary cell culture or an immortalized cell line, or may be derived from suspensions of single-celled organisms.
  • a polynucleotide encoding for a dominant negative form of a MMR protein can be introduced into the genome of an animal by producing a transgenic animal.
  • the animal can be any species for which suitable techniques are available to produce transgenic animals.
  • transgenic animals can be prepared from domestic livestock, e.g., bovine, swine, sheep, goats, horses, etc.; from animals used for the production of recombinant proteins, e.g., bovine, swine, or goats that express a recombinant polypeptide in their milk; or experimental animals for research or product testing, e.g., mice, rats, guinea pigs, hamsters, rabbits, etc.
  • Cell lines that are determined to be MMR defective can then be used as a source for producing genetically altered immunoglobulin genes in vitro by introducing whole, intact immunoglobulin genes and/or chimeric genes encoding for single chain antibodies into MMR defective cells from any tissue of the MMR defective animal.
  • a transfected cell line or a colony of transgenic animals can be used to generate new mutations in one or more gene(s) of interest.
  • a gene of interest can be any gene naturally possessed by the cell line or transgenic animal or introduced into the cell line or transgenic animal.
  • An advantage of using such cells or animals to induce mutations is that the cell or animal need not be exposed to mutagenic chemicals or radiation, which may have secondary harmful effects, both on the object of the exposure and on the workers.
  • chemical mutagens may be used in combination with MMR deficiency, which renders such mutagens less toxic due to an undetermined mechanism.
  • Hypermutable animals can then be bred and selected for those producing genetically variable B-cells that may be isolated and cloned to identify new cell lines that are useful for producing genetically variable cells.
  • the dominant negative MMR gene allele can be removed by directly knocking out the allele by technologies used by those skilled in the art or by breeding to mates lacking the dominant negative allele to select for offspring with a desired trait and a stable genome.
  • Another alternative is to use a CRE-LOX expression system, whereby the dominant negative allele is spliced from the animal genome once an animal containing a genetically diverse immunoglobulin profile has been established.
  • inducible vectors such as the steroid induced pIND (Invitrogen) or pMAM (Clonetech) vectors which express exogenous genes in the presence of corticosteroids.
  • Mutations can be detected by analyzing for alterations in the genotype of the cells or animals, for example by examining the sequence of genomic DNA, cDNA, messenger RNA, or amino acids associated with the gene of interest. Mutations can also be detected by screening for the production of antibody titers.
  • a mutant polypeptide can be detected by identifying alterations in electrophoretic mobility, spectroscopic properties, or other physical or structural characteristics of a protein encoded by a mutant gene.
  • One can also screen for altered function of the protein in situ, in isolated form, or in model systems.
  • One can screen for alteration of any property of the cell or animal associated with the function of the gene of interest, such as but not limited to lg secretion.
  • nucleic acid sequences encoding mismatch repair proteins include, but are not limited to the following: mouse PMS2 (SEQ ID NO:6); human PMS2 (SEQ ID NO:8); human PMSl (SEQ ID NO: 10) human MSH2 (SEQ ID NO:12); human MLHl (SEQ ID NO:14); and human PMS2-134 (SEQ ID NO: 16).
  • the corresponding amino acid sequences are: mouse PMS2 (SEQ ID NO:5); human PMS2 (SEQ ID NO:7); human PMSl (SEQ ID NO:9) human SH. (SEQ ID NO:ll); human MLHl (SEQ ID NO:13); and human PMS2-134 (SEQ ID NO: 15).
  • the invention provides for a method to increase the affinity of antibodies comprising replacing amino acids of the variable domain heavy and/or light chain with proline or hydroxyproline (collectively referred to as "proline").
  • proline or hydroxyproline
  • the substitution of prolines is in the heavy chain variable domain
  • the substitution of prolines is in the light chain variable domain.
  • the substitution of proline is in both the heavy chain and the light chain of the variable domain of the immunoglobulin molecule.
  • the proline substitutes for another amino acid having a non-polar sidechain (e.g., glycine, alanine, valine, leucine, isoleucine, phenylalanine, methionine, tryptophan and cysteine).
  • a non-polar sidechain e.g., glycine, alanine, valine, leucine, isoleucine, phenylalanine, methionine, tryptophan and cysteine.
  • further exchanges of amino acids having non-polar sidechains with other amino acids having non- polar sidechains may also confer increased affinity of the antibody for the antigen.
  • the amino acid substitutions are in a framework region of the heavy chain. In other embodiments, the amino acid substitutions are in a framework region of the light chain.
  • the amino acid substitutions are in a framework region of both the heavy and light chain, hi some embodiments, the amino acid substitutions are in the first framework region (FR1) of the heavy chain. In other embodiments, the amino acid substitution is in the second framework region (FR2) of the heavy chain. In other embodiments, the amino acid substitution is in the third framework region (FR3) of the heavy chain. In other embodiments, the amino acid substitution is in the fourth framework region (FR4) of the heavy chain. In some embodiments, the amino acid substitutions are in the first framework region (FR1) of the light chain. In other embodiments, the amino acid substitution is in the second framework region (FR2) of the light chain. In other embodiments, the amino acid substitution is in the third framework region (FR3) of the light chain.
  • the amino acid substitution is in the fourth framework region (FR4) of the light chain.
  • a proline substitutes for an alanine at position 6 of SEQ ID NO: 18.
  • proline substitutes for alanine at position 6 of SEQ ID NO: 18 and the glycine at position 9 of SEQ ID NO: 18, and/or the lysine at position 10 of SEQ ID NO: 18 is substituted with an amino acid having a non-polar side chain (preferably, valine and arginine, respectively).
  • proline substitutes for leucine at position 22 of SEQ ID NO:21.
  • This patent application teaches of the use of dominant negative MMR genes in antibody-producing cells, including but not limited to rodent hybridomas, human hybridomas, chimeric rodent cells producing human immunoglobulin gene products, human cells expressing immunoglobulin genes, mammalian cells producing single chain antibodies, and prokaryotic cells producing mammalian immunoglobulin genes or chimeric immunoglobulin molecules such as those contained within single-chain antibodies.
  • the cell expression systems described above that are used to produce antibodies are well known by those skilled in the art of antibody therapeutics.
  • the MMR proficient mouse H36 hybridoma cell line was transfected with various hPMS2 expression plasmids plus reporter constructs for assessing MMR activity.
  • the MMR genes were cloned into the pEF expression vector, which contains the elongation factor promoter upstream of the clomng site followed by a mammalian polyadenylation signal.
  • This vector also contains the NEOr gene that allows for selection of cells retaining this plasmid.
  • telomeres were transfected with 1 ⁇ g of each vector using polyliposomes following the manufacturer's protocol (Life Technologies). Cells were then selected in 0.5 mg/ml of G418 for 10 days and G418 resistant cells were pooled together to analyze for gene expression.
  • the pEF construct contains an intron that separates the exon 1 of the EF gene from exon 2, which is juxtaposed to the 5' end of the polylinker cloning site. This allows for a rapid reverse transcriptase polymerase chain reaction (RT-PCR) screen for cells expressing the spliced products.
  • RT-PCR reverse transcriptase polymerase chain reaction
  • RNAs were reverse transcribed using Superscript II (Life Technologies) and PCR amplified using a sense primer located in exon 1 of the EF gene (5'-ttt cgc aac ggg ttt gcc g-3') (SEQ ID NO:23) and an antisense primer (5'-gtt tea gag tta age ctt cg-3') (SEQ ID NO:24) centered at nt 283 of the published human PMS2 cDNA, which will detect both the full length as well as the PMSl 34 gene expression.
  • EXAMPLE 2 hPMS134 causes a Defect in MMR Activity and hypermutability in hybridoma cells
  • MI microsatellite instability
  • a method used to detect MMR deficiency in eukaryotic cells is to employ a reporter gene that has a polynucleotide repeat inserted within the coding region that disrupts its reading frame due to a frame shift.
  • the reporter gene will acquire random mutations (i.e. insertions and/or deletions) within the polynucleotide repeat yielding clones that contain a reporter with an open reading frame.
  • the reporter construct used the pCAR-OF, which contains a hygromycin resistance (HYG) gene plus a ⁇ -galactosidase gene containing a 29 bp out-of-frame poly-CA tract at the 5' end of its coding region.
  • the pCAR-OF reporter would not generate ⁇ -galactosidase activity unless a frame-restoring mutation (i.e., insertion or deletion) arose following transfection.
  • HBvec, HBPMS2, and HB 134 cells were each transfected with pCAR-OF vector in duplicate reactions following the protocol described in Example 1.
  • Cells were selected in 0.5 mg/ml G418 and 0.5mg/ml HYG to select for cells retaining both the MMR effector and the pCAR-OF reporter plasmids. All cultures transfected with the pCAR vector resulted in a similar number of HYG/G418 resistant cells. Cultures were then expanded and tested for ⁇ -galactosidase activity in situ as well as by biochemical analysis of cell extracts. For in.
  • Table 1 shows the results from these studies. While no ⁇ -galactosidase positive cells were observed in HBvec cells, 10% of the cells per field were ⁇ -galactosidase positive in HB134 cultures and 2% of the cells per field were ⁇ -galactosidase positive in HBPMS2 cultures.
  • Figure 2 shows the ⁇ -galactosidase activity in extracts from the various cell lines. As shown, the HB134 cells produced the highest amount of ⁇ -galactosidase, while no activity was found in the HBvec cells containing the pCAR-OF. These data demonstrate the ability to generate MMR defective hybridoma cells using dominant negative MMR gene alleles.
  • Cells were transfected with the pCAR-OF ⁇ -galactosidase reporter plasmid.
  • Transfected cells were selected in hygromycin and G418, expanded and stained with X-gal solution to measure for ⁇ -galactosidase activity (blue colored cells). 3 fields of 200 cells each were analyzed by microscopy. The results below represent the mean +/- standard deviation of these experiments.
  • EXAMPLE 3 Screening strategy to identify hybridoma clones producing antibodies with higher binding affinities and/or increased immunoglobulin production.
  • An application of the methods presented within this document is the use of MMR deficient hybridomas or other immunoglobulin producing cells to create genetic alterations within an immunoglobulin gene that will yield antibodies with altered biochemical properties.
  • An illustration of this application is demonstrated within this example whereby the HB 134 hybridoma (see Example 1), which is a MMR-defective cell line that produces an anti-human immunoglobulin type E (hlgE) MAb, is grown for 20 generations and clones are isolated in 96-well plates and screened for hlgE binding.
  • hlgE anti-human immunoglobulin type E
  • Figure 3 outlines the screening procedure to identify clones that produce high affinity MAbs, which is presumed to be due to an alteration within the light or heavy chain variable region of the protein.
  • the assay employs the use of a plate Enzyme Linked hnmunosorbant Assay (ELISA) to screen for clones that produce high- affinity MAbs.
  • ELISA plate Enzyme Linked hnmunosorbant Assay
  • plates are screened using a hlgE plate ELISA, whereby a 96 well plate is coated with 50 ⁇ ls of a 1 ⁇ g/ml hlgE solution for 4 hours at 4°C Plates are washed 3 times in calcium and magnesium free phosphate buffered saline solution (PBS " ' " ) and blocked in lOO ⁇ ls of PBS " ' " with 5% dry milk for 1 hour at room temperature. Wells are rinsed and incubated with 100 ⁇ ls of a PBS solution containing a 1:5 dilution of conditioned medium from each cell clone for 2 hours.
  • PBS calcium and magnesium free phosphate buffered saline solution
  • Plates are then washed 3 times with PBS " ' “ and incubated for 1 hour at room temperature with 50 ⁇ ls of a PBS " ' " solution containing 1:3000 dilution of a sheep anti-mouse horse radish peroxidase (HRP) conjugated secondary antibody. Plates are then washed 3 times with PBS " ' “ and incubated with 50 ⁇ ls of TMB-HRP substrate (BioRad) for 15 minutes at room temperature to detect amount of antibody produced by each clone. Reactions are stopped by adding 50 ⁇ ls of 500mM sodium bicarbonate and analyzed by OD at 415nm using a BioRad plate reader.
  • HRP horse radish peroxidase
  • Clones exhibiting an enhanced signal over background cells are then isolated and expanded into 10 ml cultures for additional characterization and confirmation of ELISA data in triplicate experiments.
  • ELISAs are also performed on conditioned medium (CM) from the same clones to measure total lg production within the conditioned medium of each well.
  • Clones that produce an increased ELISA signal and have increased antibody levels are then further analyzed for variants that over-express and/or over- secrete antibodies as described in Example 4.
  • Analysis of five 96-well plates each from HBvec or HB134 cells have found that a significant number of clones with a higher Optical Density (OD) value is observed in the MMR-defective HB134 cells as compared to the HBvec controls.
  • OD Optical Density
  • Figure 4 shows a representative example of HB134 clones producing antibodies that bind to specific antigen (in this case IgE) with a higher affinity.
  • Figure 4 provides raw data from the analysis of 96 wells of HBvec (left graph) or HB134 (right graph) which shows 2 clones from the HB134 plate to have a higher OD reading due to 1) genetic alteration of the antibody variable domain that leads to an increased binding to IgE antigen, or 2) genetic alteration of a cell host that leads to over-production/secretion of the antibody molecule.
  • Anti-Ig ELISA found that the two clones, shown in Figure 4 have lg levels within their CM similar to the surrounding wells exhibiting ower OD values.
  • Clones that produced higher OD values as determined by ELISA were further analyzed at the genetic level to confirm that mutations within the light or heavy chain variable region have occurred that lead to a higher binding affinity hence yielding to a stronger ELISA signal. Briefly, 100,000 cells are harvested and extracted for RNA using the Triazol method as described above. RNAs are reverse transcribed using Superscript II as suggested by the manufacturer (Life Technology) and PCR amplified for the antigen binding sites contained within the variable light and heavy chains. Because of the heterogeneous nature of these genes, the following degenerate primers are used to amplify light and heavy chain alleles from the parent H36 strain.
  • Light chain antisense 5'-ACT GGA TGG TGG GAA GAT GGA-3' (SEQ ID NO:2)
  • Heavy chain sense 5'-A(G/T) GTN (A/QAG CTN CAG (C/G)AG TC-3' (SEQ ID NO:3)
  • Heavy chain antisense 5'-TNC CTT G(A/G)C CCC AGT A(G/A)(A T)C-3' (SEQ ID NO:4)
  • PCR reactions using degenerate oligonucleotides are carried out at 94°C for 30 sec, 52°C for 1 min, and 72°C for 1 min for 35 cycles. Products are analyzed on agarose gels. Products of the expected molecular weights are purified from the gels by Gene Clean (Bio 101), cloned into T-tailed vectors, and sequenced to identify the wild type sequence of the variable light and heavy chains. Once the wild type sequence has been determined, non- degenerate primers were made for RT-PCR amplification of positive HB134 clones. Both the light and heavy chains were amplified, gel purified and sequenced using the corresponding sense and antisense primers.
  • RT-PCR products give representative sequence data of the endogenous immunoglobulin gene and not due to PCR induced mutations. Sequences from clones were then compared to the wild type sequence for sequence comparison.
  • An example of the ability to create in vivo mutations within an immunoglobulin light or heavy chain is shown in Figure 5, where HB 134 clone92 was identified by ELISA to have an increased signal for hlgE.
  • the light chain was amplified using specific sense, and antisense primers.
  • the light chain was RT-PCR amplified and the resulting product was purified and analyzed on an automated ABI377 sequencer.
  • a residue -4 upstream of the CDR region 3 had a genetic change from ACT to TCT, which results in a Thr to Ser change within the framework region just preceding the CDR#3.
  • a residue -6 upstream of the CDR region had a genetic change from CCC to CTC, which results in a Pro to His change within framework region preceding CDR#2.
  • a common method for producing humanized antibodies is to graft CDR sequences from a MAb (produced by immunizing a rodent host) onto a human lg backbone, and transfection of the chimeric genes into Chinese Hamster Ovary (CHO) cells whih in turn produce a functional Ab that is secreted by the CHO cells (Shields, R.L., et al. (1995) Anti-IgE monoclonal antibodies that inhibit allergen-specific histamine release. Int. Arch. Allergy Immunol. 107:412-413).
  • the methods described within this application are also useful for generating genetic alterations within lg genes or chimeric Igs transfected within host cells such as rodent cell lines, plants, yeast and prokaryotes (Frigerio L, et al. (2000) Assembly, secretion, and vacuolar delivery of a hybrid immunoglobulin in plants. Plant Physiol 123 : 1483-1494). [0066] These data demonstrate the ability to generate hypermutable hybridomas, or other lg producing host cells that can be grown and selected, to identify structurally altered immunoglobulins yielding antibodies with enhanced biochemical properties, including but not limited to increased antigen binding affinity.
  • hypermutable clones that contain missense mutations, within the immunoglobulin gene that result in an amino acid change or changes can be then further characterized for in vivo stability, antigen clearance, on-off binding to antigens, etc. Clones can also be further expanded for subsequent rounds of in vivo mutations and can be screened using the strategy listed above. [0067] The use of chemical mutagens to produce genetic mutations in cells or whole organisms are limited due to the toxic effects that these agents have on "normal" cells.
  • Example 4 Generation of antibody producing cells with enhanced antibody production
  • HB134 clones were assayed by ELISA for elevated lg levels. Analysis of 480 clones showed that a significant number of clones had elevated MAb product levels in their CM. Quantification showed that several of these clones produced greater than 500ngs/ml of MAb due to either enhanced expression and/or secretion as compared to clones from the H36 cell line.
  • CM conditioned medium
  • Figure 6 shows a representative analysis where a subset of clones had enhanced lg production which accounted for increased Ab production (Lane 5) while others had a similar steady state level as the control sample, yet had higher levels of Ab within the CM.
  • This strategy allows for the use of chemical mutagens to be used in MMR-defective Ab producing cells as a method for increasing additional mutations within immunoglobulin genes or chimeras that may yield functional Abs with altered biochemical properties such as enhanced binding affinity to antigen, etc.
  • Example 5 Establishment of genetic stability in hybridoma cells with new output trait.
  • the initial steps of MMR are dependent on two protein complexes, called MutS ⁇ and MutL ⁇ (Nicolaides et al. (1998) A Naturally Occurring hPMS2 Mutation Can Confer a Dominant Negative Mutator Phenotype. Mol. Cell. Biol. 18:1635-1641).
  • Dominant negative MMR alleles are able to perturb the formation of these complexes with downstream biochemicals involved in the excision and polymerization of nucleotides comprising the "corrected" nucleotides.
  • Examples from this application show the ability of a truncated MMR allele (PMSl 34) as well as a full length human PMS2 when expressed in a hybridoma cell line is capable of blocking MMR resulting in a hypermutable cell line that gains genetic alterations throughout its entire genome per cell division.
  • a cell line is produced that contains genetic alterations within genes encoding for an antibody, a single chain antibody, over expression of immunoglobulin genes and/or enhanced secretion of antibody, it is desirable to restore the genomic integrity of the cell host. This can be achieved by the use of inducible vectors whereby dominant negative MMR genes are cloned into such vectors, introduced into Ab producing cells and the cells are cultured in the presence of inducer molecules and/or conditions.
  • Inducible vectors include but are not limited to chemical regulated promoters such as the steroid inducible MMTV, tetracycline regulated promoters, temperature sensitive MMR gene alleles, and temperature sensitive promoters.
  • chemical regulated promoters such as the steroid inducible MMTV, tetracycline regulated promoters, temperature sensitive MMR gene alleles, and temperature sensitive promoters.
  • Cisplatin and adriamycin resistance are associated with MutLa and mismatch repair deficiency in an ovarian tumor cell line. J. Biol. Chem. 271:9645-19648). It is known from previous studies in both prokaryotes and eukaryotes that the separate enzymatic components mediate repair from the two different directions. Our results, in combination with those of Drummond et al. (Shields, R.L., et al. (1995) Anti-IgE monoclonal antibodies that inhibit allergen-specific histamine release. Int. Arch Allergy Immunol.
  • Blockade of MMR in such cells can be through the use of dominant negative MMR gene alleles from any species including bacteria, yeast, protozoa, insects, rodents, primates, mammalian cells, and man.
  • Blockade of MMR can also be generated through the use of antisense RNA or deoxynucleotides directed to any of the genes involved in the MMR biochemical pathway.
  • Blockade of MMR can be through the use of polypeptides that interfere with subunits of the MMR complex including but not limited to antibodies.
  • the blockade of MMR may be through the use chemicals such as but not limited to nonhydrolyzable ATP analogs, which have been shown to block MMR (Galio, L, et al. (1999) ATP hydrolysis-dependent formation of a dynamic ternary nucleoprotein complex with MutS and MutL. Nucl Acids Res. 27:2325-23231).
  • the genetically altered antibodies show the following sequence differences and consensus sequence:
  • the data shows that for the light chain, a substitution in the second framework region (FR2) of the light chain at position 22 of SEQ TD NO:21 to a proline increased the binding affinity of the antibody.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

On peut utiliser des allèles dominantes négatives de gènes humains de réparation des désappariements pour produire des cellules et organismes hypermutables. En introduisant ces gènes dans des animaux transgéniques, on peut préparer de nouvelles lignées de cellules et de nouvelles variétés d'animaux plus efficacement qu'en se basant sur le taux naturel de mutation. Ces méthodes, qui s'avèrent utiles pour créer une diversité génétique parmi des gènes d'immunoglobine dirigés contre des antigènes d'intérêt pour produire des anticorps modifiés à activité biochimique accrue, le sont de plus pour créer des cellules produisant à fort débit des d'anticorps. L'invention porte également sur des méthodes accroissant l'affinité d'anticorps monoclonaux, et sur des anticorps monoclonaux à affinité accrue.
PCT/US2003/028722 2002-09-13 2003-09-12 Anticorps et methodes de creation d'anticorps genetiquement modifies a forte affinite WO2004024871A2 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002544124A CA2544124A1 (fr) 2002-09-13 2003-09-12 Anticorps et methodes de creation d'anticorps genetiquement modifies a forte affinite
EP03754530A EP1556508A4 (fr) 2002-09-13 2003-09-12 Anticorps et methodes de creation d'anticorps genetiquement modifies a forte affinite
AU2003272352A AU2003272352A1 (en) 2002-09-13 2003-09-12 Antibodies and methods for generating genetically altered antibodies with high affinity
JP2004536226A JP2006503035A (ja) 2002-09-13 2003-09-12 抗体および高親和性を有する遺伝的に改変された抗体の生成法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/243,130 2002-09-13
US10/243,130 US7235643B2 (en) 2000-11-07 2002-09-13 Antibodies and methods for generating genetically altered antibodies with high affinity

Publications (2)

Publication Number Publication Date
WO2004024871A2 true WO2004024871A2 (fr) 2004-03-25
WO2004024871A3 WO2004024871A3 (fr) 2005-05-19

Family

ID=31991557

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/028722 WO2004024871A2 (fr) 2002-09-13 2003-09-12 Anticorps et methodes de creation d'anticorps genetiquement modifies a forte affinite

Country Status (6)

Country Link
US (2) US7235643B2 (fr)
EP (1) EP1556508A4 (fr)
JP (1) JP2006503035A (fr)
AU (1) AU2003272352A1 (fr)
CA (1) CA2544124A1 (fr)
WO (1) WO2004024871A2 (fr)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005011735A1 (fr) * 2003-07-29 2005-02-10 Morphotek, Inc. Anticorps et procedes permettant de produire des anticorps genetiquement modifies presentant une fonction effectrice amelioree
EP1572971A2 (fr) * 2002-11-15 2005-09-14 Morphotek Inc. Methodes de generation d'une quantite elevee d'anticorps a partir d'hybridomes crees par immunisation in vitro
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US7671179B2 (en) 2000-11-07 2010-03-02 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with high affinity
US7759464B2 (en) 2004-07-14 2010-07-20 Greenovation Biotech Gmbh N-glycosylated antibody
US7973136B2 (en) 2005-10-06 2011-07-05 Xencor, Inc. Optimized anti-CD30 antibodies
US8039592B2 (en) 2002-09-27 2011-10-18 Xencor, Inc. Optimized Fc variants and methods for their generation
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US8124731B2 (en) 2002-03-01 2012-02-28 Xencor, Inc. Optimized Fc variants and methods for their generation
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US8318907B2 (en) 2004-11-12 2012-11-27 Xencor, Inc. Fc variants with altered binding to FcRn
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US8394374B2 (en) 2006-09-18 2013-03-12 Xencor, Inc. Optimized antibodies that target HM1.24
US8524867B2 (en) 2006-08-14 2013-09-03 Xencor, Inc. Optimized antibodies that target CD19
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US9040041B2 (en) 2005-10-03 2015-05-26 Xencor, Inc. Modified FC molecules
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
US9200079B2 (en) 2004-11-12 2015-12-01 Xencor, Inc. Fc variants with altered binding to FcRn
US9657106B2 (en) 2003-03-03 2017-05-23 Xencor, Inc. Optimized Fc variants
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants
US11820830B2 (en) 2004-07-20 2023-11-21 Xencor, Inc. Optimized Fc variants
US11932685B2 (en) 2007-10-31 2024-03-19 Xencor, Inc. Fc variants with altered binding to FcRn

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080260731A1 (en) * 2002-03-01 2008-10-23 Bernett Matthew J Optimized antibodies that target cd19
US20080254027A1 (en) * 2002-03-01 2008-10-16 Bernett Matthew J Optimized CD5 antibodies and methods of using the same
US20070148171A1 (en) * 2002-09-27 2007-06-28 Xencor, Inc. Optimized anti-CD30 antibodies
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
US20070275460A1 (en) * 2003-03-03 2007-11-29 Xencor.Inc. Fc Variants With Optimized Fc Receptor Binding Properties
US20050106222A1 (en) * 2003-06-11 2005-05-19 Ailor Eric N. Method to increase protein production in culture
EP1697520A2 (fr) * 2003-12-22 2006-09-06 Xencor, Inc. Polypeptides fc a nouveaux sites de liaison de ligands fc
EP1737890A2 (fr) * 2004-03-24 2007-01-03 Xencor, Inc. Variantes d'immunoglobuline a l'exterieur de la region fc
US20060074225A1 (en) * 2004-09-14 2006-04-06 Xencor, Inc. Monomeric immunoglobulin Fc domains
WO2006076594A2 (fr) * 2005-01-12 2006-07-20 Xencor, Inc. Anticorps et proteines de fusion fc a immunogenicite modifiee
JP2009504685A (ja) * 2005-08-15 2009-02-05 アラーナ・テラピューティクス・リミテッド 新世界霊長類フレームワーク領域を用いる設計された抗体
ZA200802247B (en) * 2005-08-15 2009-08-26 Arana Therapeutics Ltd Chimeric antibodies with new world primate regions
US20090286962A1 (en) * 2005-12-20 2009-11-19 Woolven Benjamin P Chimeric antibodies with part new world primate binding regions
NZ569988A (en) * 2006-02-01 2011-09-30 Cephalon Australia Pty Ltd Domain antibody construct which binds to human TNF-alpha and contains a modified hinge region sequence and a truncated CH1 domain
AU2007237985A1 (en) * 2006-04-17 2007-10-25 Morphotek, Inc. Whole genome evolution technology applied to improve protein and antibody yields by cells
US20080139790A1 (en) * 2006-12-08 2008-06-12 Jennings Philip A Chimeric antibodies
EP2115127A4 (fr) * 2007-01-17 2010-04-07 Life Technologies Corp Procédés et compositions pour améliorer la santé de cellules en culture
BRPI0807952A2 (pt) 2007-02-20 2014-06-10 Anaptysbio Inc Sistemas de hipermutação somática
US7580304B2 (en) * 2007-06-15 2009-08-25 United Memories, Inc. Multiple bus charge sharing
LT2954779T (lt) 2009-12-10 2019-05-27 Regeneron Pharmaceuticals, Inc. Pelės, gaminančios sunkiosios grandinės antikūnus
WO2011091078A2 (fr) 2010-01-19 2011-07-28 Xencor, Inc. Variants d'anticorps possédant une activité complémentaire accrue

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO2002037967A1 (fr) 2000-11-07 2002-05-16 Morphotek Inc. Procedes pour generer des lignees cellulaires produisant des anticorps genetiquement modifies presentant des caracteristiques ameliorees
US20030143682A1 (en) 2000-11-07 2003-07-31 Nicolaides Nicholas C. Antibodies and methods for generating genetically altered antibodies with high affinity
WO2005011735A1 (fr) 2003-07-29 2005-02-10 Morphotek, Inc. Anticorps et procedes permettant de produire des anticorps genetiquement modifies presentant une fonction effectrice amelioree

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06327479A (ja) * 1993-05-20 1994-11-29 Dai Ichi Pure Chem Co Ltd 抗チミンダイマー抗体tdm−2可変領域のdna配列
US6191268B1 (en) * 1993-12-17 2001-02-20 Dana-Farber Cancer Institute Compositions and methods relating to DNA mismatch repair genes
CA2263958C (fr) 1995-07-26 2012-06-19 Mixis France Recombinaison homologue dans des cellules eucaryotes a systeme de reparation des mesappariements inactive
US5907079A (en) * 1996-01-18 1999-05-25 Amgen Canada Inc. MSH2 disrupted mice develop lymphomas
US5885827A (en) * 1996-01-23 1999-03-23 The Regents Of The Universtiy Of California Eukaryotic high rate mutagenesis system
US6326204B1 (en) * 1997-01-17 2001-12-04 Maxygen, Inc. Evolution of whole cells and organisms by recursive sequence recombination
AUPO974597A0 (en) 1997-10-10 1997-11-06 Rhone-Poulenc Agro Methods for obtaining plant varieties
US6146894A (en) 1998-04-14 2000-11-14 The Johns Hopkins University Method for generating hypermutable organisms
US6808894B1 (en) * 2000-11-07 2004-10-26 Morphotek, Inc. Methods for generating genetically altered antibody producing cell lines with improved antibody characteristics
ATE501635T1 (de) 2001-01-15 2011-04-15 Morphotek Inc Chemische hemmstoffe des mismatch-repair
US20040002500A1 (en) 2002-06-28 2004-01-01 Fabre Kramer Pharmaceutical, Inc. Methods for treating attention deficit disorder

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO2002037967A1 (fr) 2000-11-07 2002-05-16 Morphotek Inc. Procedes pour generer des lignees cellulaires produisant des anticorps genetiquement modifies presentant des caracteristiques ameliorees
US20030143682A1 (en) 2000-11-07 2003-07-31 Nicolaides Nicholas C. Antibodies and methods for generating genetically altered antibodies with high affinity
WO2005011735A1 (fr) 2003-07-29 2005-02-10 Morphotek, Inc. Anticorps et procedes permettant de produire des anticorps genetiquement modifies presentant une fonction effectrice amelioree

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
BIGNAMI M: "Unmasking a killer: DNA O(6)-n-methylguanine and the cytotoxicity of methylating agents", MUTAT. RES., vol. 462, 2000, pages 71 - 82
DRUMMOND, J.T. ET AL.: "Cisplatin and adriamycin resistance are associated with MutLa and mismatch repair deficiency in an ovarian tumor cell line", J. BIOL. CHEM., vol. 271, 1996, pages 9645 - 19648
EMERY, S.C.; W.J. HARRIS: "ANTIBODY ENGINEERING C.A.K.", 1995, OXFORD UNIVERSITY PRESS, article "Strategies for humanizing antibodies", pages: 159 - 183
ESHLEMAN J.R.; S.D. MARKOWITZ: "Mismatch repair defects in human carcinogenesis", HUM. MOL. GENET., vol. 5, 1996, pages 1489 - 494
FIEDLER, U.; CONRAD, U.: "High-level production and long-term storage of engineered antibodies in transgeilic tobacco seeds", BIOL/TECHNOLOGY, vol. 13, 1995, pages 1090 - 1093
FRIGERIO L. ET AL.: "Assembly, secretion, and vacuolar delivery of a hybrid immunoglobulin in plants", PLANT PHYSIOL., vol. 123, 2000, pages 1483 - 1494
GALIO, L. ET AL.: "ATP hydrolysis-dependent formation of a dynamic ternary nucleoprotein complex with MutS and MutL", NUCL. ACIDS RES., vol. 27, 1999, pages 2325 - 23231
GLASER, V.: "Can ReoPro repolish tarnished monoclonal therapeutics?", NAT. BIOTECHNOL., vol. 14, 1996, pages 1216 - 1217
GLASER, V.: "Can ReoPro repolish tarnished monoclonal therapeutics?", NAT. BIOTECHOL., vol. 14, 1996, pages 1216 - 1217
KHAZAELI, M.B. ET AL.: "Human immune response to monoclonal antibodies", J IMMUNOTHER., vol. 15, 1994, pages 42 - 52
LIU, T. ET AL.: "Microsatellite instability as a predictor of a mutation in a DNA mismatch repair gene in familial colorectal cancer", GENES CHROMOSOMES CANCER, vol. 27, 2000, pages 17 - 25
NEUBERGER, M.; GRUGGERMANN, M.: "Monoclonal antibodies. Mice perform a human repertoire", NATURE, vol. 386, 1997, pages 25 - 26
NEUBERGER, M.; M. GRUGGEMANN: "Monoclonal antibodies. Mice perfonn a human repertoire", NATURE, vol. 386, 1997, pages 25 - 26
NICOLAIDES N.C.; K.W. KINZLER; B. VOGELSTEIN: "Analysis of the 5' region of PMS2 reveals heterogenous transcripts and a novel overlapping gene", GENOMICS, vol. 29, 1995, pages 329 - 334
NICOLAIDES, N.C. ET AL.: "Genomic organization of the human PMS2 gene family", GENOMICS, vol. 30, 1995, pages 195 - 206
NICOLAIDES, N.C. ET AL.: "The Jun family members, c-JUN and JUND, transactivate the human c-myb promoter via an Apl like element", J. BIOL. CHEM., vol. 267, 1992, pages 19665 - 19672
PALOMBO, F. ET AL.: "Mismatch repair and cancer", NATURE, vol. 36, 1994, pages 417
REFF, M.E.: "High-level production of recombinant immunoglobulins in mammalian cells", CURR. OPIN. BIOTECHMOL., vol. 4, 1993, pages 573 - 576
REFF, M.E.: "High-level production of recombinant immunoglobulins in mammalian cells", CURR. OPIN. BIOTECHNOL., vol. 4, 1993, pages 573 - 576
SAEZ-LLORENS, X.E. ET AL.: "Safety and pharmacokinetics of an intramuscular humanized monoclonal antibody to respiratory syncytial virus in premature infants and infants with bronchopulmonary dysplasia", PEDIAT. INTECT. DIS. J., vol. 17, 1998, pages 787 - 791
See also references of EP1556508A4
SHIELD, C.F. ET AL.: "A cost-effective analysis of OKT3 induction therapy in cadaveric kidney transplantation", AM. J. KIDNEY DIS., vol. 27, 1996, pages 855 - 864
SHIELDS, R.L. ET AL.: "Anti-IgE monoclonal antibodies that inhibit allergen- specific histamine release", INT. ARCH. ALLERGY IMMUNOL., vol. 107, 1995, pages 412 - 413
WEINER, L.M.: "Monoclonal antibody therapy of cancer", SEMIN. ONCOL., vol. 26, 1999, pages 43 - 51
WEMER, L.M.: "Monoclonal antibody therapy of cancer", SEMIN. ONCOL., vol. 26, 1999, pages 43 - 51

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7671179B2 (en) 2000-11-07 2010-03-02 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with high affinity
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US8734791B2 (en) 2002-03-01 2014-05-27 Xencor, Inc. Optimized fc variants and methods for their generation
US8124731B2 (en) 2002-03-01 2012-02-28 Xencor, Inc. Optimized Fc variants and methods for their generation
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US8039592B2 (en) 2002-09-27 2011-10-18 Xencor, Inc. Optimized Fc variants and methods for their generation
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US10183999B2 (en) 2002-09-27 2019-01-22 Xencor, Inc. Optimized Fc variants and methods for their generation
US8809503B2 (en) 2002-09-27 2014-08-19 Xencor, Inc. Optimized Fc variants and methods for their generation
US9353187B2 (en) 2002-09-27 2016-05-31 Xencor, Inc. Optimized FC variants and methods for their generation
US8383109B2 (en) 2002-09-27 2013-02-26 Xencor, Inc. Optimized Fc variants and methods for their generation
US9193798B2 (en) 2002-09-27 2015-11-24 Xencor, Inc. Optimized Fc variants and methods for their generation
US10184000B2 (en) 2002-09-27 2019-01-22 Xencor, Inc. Optimized Fc variants and methods for their generation
US8093359B2 (en) 2002-09-27 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US8858937B2 (en) 2002-09-27 2014-10-14 Xencor, Inc. Optimized Fc variants and methods for their generation
US7754450B2 (en) 2002-11-15 2010-07-13 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
EP1572971B1 (fr) * 2002-11-15 2009-09-30 Morphotek Inc. Methodes de generation d'une quantite elevee d'anticorps a partir d'hybridomes crees par immunisation in vitro
AU2003295576B2 (en) * 2002-11-15 2011-03-17 Eisai, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
EP1572971A2 (fr) * 2002-11-15 2005-09-14 Morphotek Inc. Methodes de generation d'une quantite elevee d'anticorps a partir d'hybridomes crees par immunisation in vitro
US8445229B2 (en) 2002-11-15 2013-05-21 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
US8735545B2 (en) 2003-03-03 2014-05-27 Xencor, Inc. Fc variants having increased affinity for fcyrllc
US10113001B2 (en) 2003-03-03 2018-10-30 Xencor, Inc. Fc variants with increased affinity for FcyRIIc
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US10584176B2 (en) 2003-03-03 2020-03-10 Xencor, Inc. Fc variants with increased affinity for FcγRIIc
US9657106B2 (en) 2003-03-03 2017-05-23 Xencor, Inc. Optimized Fc variants
US9663582B2 (en) 2003-03-03 2017-05-30 Xencor, Inc. Optimized Fc variants
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
WO2005011735A1 (fr) * 2003-07-29 2005-02-10 Morphotek, Inc. Anticorps et procedes permettant de produire des anticorps genetiquement modifies presentant une fonction effectrice amelioree
US7807416B2 (en) 2003-07-29 2010-10-05 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with enhanced effector function
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US8323650B2 (en) 2004-07-14 2012-12-04 Meridian Biopharmaceuticals Gmbh Method of treating lewis Y-expressing tumors
US7759464B2 (en) 2004-07-14 2010-07-20 Greenovation Biotech Gmbh N-glycosylated antibody
US11820830B2 (en) 2004-07-20 2023-11-21 Xencor, Inc. Optimized Fc variants
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US8324351B2 (en) 2004-11-12 2012-12-04 Xencor, Inc. Fc variants with altered binding to FcRn
US8338574B2 (en) 2004-11-12 2012-12-25 Xencor, Inc. FC variants with altered binding to FCRN
US8883973B2 (en) 2004-11-12 2014-11-11 Xencor, Inc. Fc variants with altered binding to FcRn
US9200079B2 (en) 2004-11-12 2015-12-01 Xencor, Inc. Fc variants with altered binding to FcRn
US11198739B2 (en) 2004-11-12 2021-12-14 Xencor, Inc. Fc variants with altered binding to FcRn
US8852586B2 (en) 2004-11-12 2014-10-07 Xencor, Inc. Fc variants with altered binding to FcRn
US8318907B2 (en) 2004-11-12 2012-11-27 Xencor, Inc. Fc variants with altered binding to FcRn
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US10336818B2 (en) 2004-11-12 2019-07-02 Xencor, Inc. Fc variants with altered binding to FcRn
US9803023B2 (en) 2004-11-12 2017-10-31 Xencor, Inc. Fc variants with altered binding to FcRn
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US9040041B2 (en) 2005-10-03 2015-05-26 Xencor, Inc. Modified FC molecules
US9574006B2 (en) 2005-10-06 2017-02-21 Xencor, Inc. Optimized anti-CD30 antibodies
US7973136B2 (en) 2005-10-06 2011-07-05 Xencor, Inc. Optimized anti-CD30 antibodies
US8524867B2 (en) 2006-08-14 2013-09-03 Xencor, Inc. Optimized antibodies that target CD19
US9803020B2 (en) 2006-08-14 2017-10-31 Xencor, Inc. Optimized antibodies that target CD19
US10626182B2 (en) 2006-08-14 2020-04-21 Xencor, Inc. Optimized antibodies that target CD19
US11618788B2 (en) 2006-08-14 2023-04-04 Xencor, Inc. Optimized antibodies that target CD19
US8394374B2 (en) 2006-09-18 2013-03-12 Xencor, Inc. Optimized antibodies that target HM1.24
US9040042B2 (en) 2006-09-18 2015-05-26 Xencor, Inc. Optimized antibodies that target HM1.24
US11932685B2 (en) 2007-10-31 2024-03-19 Xencor, Inc. Fc variants with altered binding to FcRn
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants

Also Published As

Publication number Publication date
EP1556508A4 (fr) 2006-10-04
WO2004024871A3 (fr) 2005-05-19
US20070244302A1 (en) 2007-10-18
US7235643B2 (en) 2007-06-26
US7671179B2 (en) 2010-03-02
JP2006503035A (ja) 2006-01-26
CA2544124A1 (fr) 2004-03-25
AU2003272352A1 (en) 2004-04-30
US20030143682A1 (en) 2003-07-31
EP1556508A2 (fr) 2005-07-27

Similar Documents

Publication Publication Date Title
WO2004024871A2 (fr) Anticorps et methodes de creation d'anticorps genetiquement modifies a forte affinite
US7807416B2 (en) Antibodies and methods for generating genetically altered antibodies with enhanced effector function
US20100021996A1 (en) Genetically Altered Anti-body Producing Cell Lines With Improved Antibody Characteristics
KR101553244B1 (ko) 중쇄 항체를 만드는 마우스
US20110185441A1 (en) Transgenic animals and methods of making recombinant antibodies
US20090155797A1 (en) Methods For Generating Enhanced Antibody-Producing Cell Lines With Improved Growth Characteristics
US11832598B2 (en) Genetically modified non-human animals and methods for producing heavy chain-only antibodies
EP1345495B1 (fr) Procedes pour generer des lignees cellulaires produisant des anticorps genetiquement modifies presentant des caracteristiques ameliorees
CN111065650A (zh) 单链VH-L1-Cκ-L2-CH1-抗体
ES2588910T3 (es) Método para generar diversidad
EP3476942B1 (fr) Gènes de dh germinaux longs et anticorps hcdr3 longs

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2544124

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004536226

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003272352

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003754530

Country of ref document: EP

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWP Wipo information: published in national office

Ref document number: 2003754530

Country of ref document: EP